Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Microbiol Spectr ; 12(8): e0016724, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38916320

RESUMEN

Mycobacterium tuberculosis (Mtb) as well as nontuberculous mycobacteria are intracellular pathogens whose treatment is extensive and increasingly impaired due to the rise of mycobacterial drug resistance. The loss of antibiotic efficacy has raised interest in the identification of host-directed therapeutics (HDT) to develop novel treatment strategies for mycobacterial infections. In this study, we identified amiodarone as a potential HDT candidate that inhibited both intracellular Mtb and Mycobacterium avium in primary human macrophages without directly impairing bacterial growth, thereby confirming that amiodarone acts in a host-mediated manner. Moreover, amiodarone induced the formation of (auto)phagosomes and enhanced autophagic targeting of mycobacteria in macrophages. The induction of autophagy by amiodarone is likely due to enhanced transcriptional regulation, as the nuclear intensity of the transcription factor EB, the master regulator of autophagy and lysosomal biogenesis, was strongly increased. Furthermore, blocking lysosomal degradation with bafilomycin impaired the host-beneficial effect of amiodarone. Finally, amiodarone induced autophagy and reduced bacterial burden in a zebrafish embryo model of tuberculosis, thereby confirming the HDT activity of amiodarone in vivo. In conclusion, we have identified amiodarone as an autophagy-inducing antimycobacterial HDT that improves host control of mycobacterial infections. IMPORTANCE: Due to the global rise in antibiotic resistance, there is a strong need for alternative treatment strategies against intracellular bacterial infections, including Mycobacterium tuberculosis (Mtb) and non-tuberculous mycobacteria. Stimulating host defense mechanisms by host-directed therapy (HDT) is a promising approach for treating mycobacterial infections. This study identified amiodarone, an antiarrhythmic agent, as a potential HDT candidate that inhibits the survival of Mtb and Mycobacterium avium in primary human macrophages. The antimycobacterial effect of amiodarone was confirmed in an in vivo tuberculosis model based on Mycobacterium marinum infection of zebrafish embryos. Furthermore, amiodarone induced autophagy and inhibition of the autophagic flux effectively impaired the host-protective effect of amiodarone, supporting that activation of the host (auto)phagolysosomal pathway is essential for the mechanism of action of amiodarone. In conclusion, we have identified amiodarone as an autophagy-inducing HDT that improves host control of a wide range of mycobacteria.


Asunto(s)
Amiodarona , Autofagia , Macrófagos , Mycobacterium tuberculosis , Tuberculosis , Pez Cebra , Amiodarona/farmacología , Autofagia/efectos de los fármacos , Animales , Pez Cebra/microbiología , Humanos , Macrófagos/microbiología , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Modelos Animales de Enfermedad , Mycobacterium avium/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/microbiología
2.
PLoS One ; 19(5): e0303516, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38728330

RESUMEN

Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.


Asunto(s)
Succinatos , Succinatos/farmacología , Succinatos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Micobacterias no Tuberculosas/efectos de los fármacos , Micobacterias no Tuberculosas/crecimiento & desarrollo , Células THP-1 , Infecciones por Mycobacterium no Tuberculosas/microbiología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/crecimiento & desarrollo , Mycobacterium abscessus/efectos de los fármacos , Mycobacterium abscessus/crecimiento & desarrollo , Mycobacterium abscessus/metabolismo
4.
Microbes Infect ; 22(10): 567-575, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32882411

RESUMEN

Mycobacterium avium complex is a causative organism for refractory diseases. In this study, we examined the effects of N-acetyl-cysteine on M. avium infection in vitro and in vivo. N-acetyl-cysteine treatment suppressed the growth of M. avium in A549 cells in a concentration-dependent manner. This effect was related to the induction of the antibacterial peptide human ß-defensin-2. In a mouse model, N-acetyl-cysteine treatment significantly reduced the number of bacteria in the lungs and induced murine ß-defensin-3. In interleukin-17-deficient mice, the effects of N-acetyl-cysteine disappeared, indicating that these mechanisms may be mediated by interleukin-17. Moreover, an additional reduction in bacterial load was observed in mice administered N-acetyl-cysteine in combination with clarithromycin. Our findings demonstrate the potent antimycobacterial effects of N-acetyl-cysteine against M. avium by inducing antimicrobial peptide, suggesting that N-acetyl-cysteine may have applications as an alternative to classical treatment regimens.


Asunto(s)
Acetilcisteína/farmacología , Antituberculosos/farmacología , Infección por Mycobacterium avium-intracellulare/tratamiento farmacológico , Mycobacterium avium/efectos de los fármacos , beta-Defensinas/metabolismo , Células A549 , Acetilcisteína/uso terapéutico , Animales , Antituberculosos/uso terapéutico , Carga Bacteriana/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Interleucina-17/metabolismo , Pulmón/efectos de los fármacos , Pulmón/microbiología , Ratones , Mycobacterium avium/crecimiento & desarrollo , Infección por Mycobacterium avium-intracellulare/metabolismo , Infección por Mycobacterium avium-intracellulare/microbiología , Células RAW 264.7 , Transducción de Señal
5.
Int J Antimicrob Agents ; 56(4): 106148, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32853674

RESUMEN

Despite early treatment with antimycobacterial combination therapy, drug resistance continues to emerge. Maintenance of redox homeostasis is essential for Mycobacterium avium (M. avium) survival and growth. The aim of the present study was to investigate the antimycobacterial activity of two pro-glutathione (pro-GSH) drugs that are able to induce redox stress in M. avium and to modulate cytokine production by macrophages. Hence, we investigated two molecules shown to possess antiviral and immunomodulatory properties: C4-GSH, an N-butanoyl GSH derivative; and I-152, a prodrug of N-acetyl-cysteine (NAC) and ß-mercaptoethylamine (MEA). Both molecules showed activity against replicating M. avium, both in the cell-free model and inside macrophages. Moreover, they were even more effective in reducing the viability of bacteria that had been kept in water for 7 days, proving to be active both against replicating and non-replicating bacteria. By regulating the macrophage redox state, I-152 modulated cytokine production. In particular, higher levels of interferon-gamma (IFN-γ), interleukin 1 beta (IL-1ß), IL-18 and IL-12, which are known to be crucial for the control of intracellular pathogens, were found after I-152 treatment. Our results show that C4-GSH and I-152, by inducing perturbation of redox equilibrium, exert bacteriostatic and bactericidal activity against M. avium. Moreover, I-152 can boost the host response by inducing the production of cytokines that serve as key regulators of the Th1 response.


Asunto(s)
Acetilcisteína/análogos & derivados , Antibacterianos/farmacología , Cisteamina/análogos & derivados , Glutatión/farmacología , Mycobacterium avium/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Acetilcisteína/farmacología , Cisteamina/farmacología , Citocinas/metabolismo , Glutatión/análogos & derivados , Humanos , Macrófagos/metabolismo , Pruebas de Sensibilidad Microbiana , Estrés Oxidativo/efectos de los fármacos
6.
J Antibiot (Tokyo) ; 73(8): 581-584, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32132675

RESUMEN

The marine actinomycete strain OPMA02852, identified as the genus Streptomyces, was found to produce anti-mycobacterial compounds against Mycobacterium avium complex (MAC). One new compound, designated as steffimycin E (1), was isolated together with three known steffimycins (steffimycin (2), 10-dihydrosteffimycin (3), and 8-demethoxysteffimycin (4)) from the culture broth of this producing microorganism by solvent extraction, ODS column chromatography, and preparative HPLC. Compound 1 has a tetracyclic quinone structure with a sugar moiety. Compound 1 exhibited anti-mycobacterial activity against M. intracellulare, M. bovis BCG, and M. smegmatis.


Asunto(s)
Antraciclinas/farmacología , Antituberculosos/farmacología , Complejo Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/efectos de los fármacos , Streptomyces/química , Antraciclinas/química , Antituberculosos/química , Línea Celular Tumoral , Células HeLa , Humanos , Infección por Mycobacterium avium-intracellulare/tratamiento farmacológico
7.
Braz. arch. biol. technol ; Braz. arch. biol. technol;63: e20190408, 2020. tab
Artículo en Inglés | LILACS | ID: biblio-1132168

RESUMEN

Abstract Propolis is a resinous substance collected and processed by Apis mellifera from parts of plants, buds and exudates. In Minas Gerais (MG) state, Brazil, green propolis is produced from the collection of resinous substance found in shoot apices of Baccharis dracunculifolia. This paper aims to investigate the chemical composition and in vitro antioxidant, anti-Helicobacter pylori, antimycobacterial and antiproliferative activities of essential oil (EO) from Brazilian green propolis (BGP-EO). The oil showed high antibacterial activity against H. pylori (MIC = 6.25 µg/mL), Mycobacterium avium (MIC = 62.5 µg/mL) and M. tuberculosis (MIC = 64 µg/mL). Its antioxidant activity was evaluated in vitro by both DPPH (IC50 = 23.48 µg/mL) and ABTS (IC50 = 32.18 µg/mL) methods. The antiproliferative activity in normal (GM07492A, lung fibroblasts) and tumor cell lines (MCF-7, HeLa and M059J) was analyzed by the XTT assay. BGP-EO showed inhibition of normal cell growth at 68.93 ± 2.56 µg/mL. Antiproliferative activity was observed against human tumor cell lines, whose IC50 values were 56.17, 66.43 and -65.83 µg/mL for MCF-7, HeLa and M059J cells, respectively. Its major constituents, which were determined by GC-FID and GC-MS, were carvacrol (20.7 %), acetophenone (13.5 %), spathulenol (11.0 %), (E)-nerolidol (9.7 %) and β-caryophyllene (6.2 %). These results showed the effectiveness of BGP-EO as a natural product which has promising biological activities.


Asunto(s)
Própolis/química , Antiinfecciosos/farmacología , Antioxidantes/farmacología , Brasil , Aceites Volátiles/uso terapéutico , Helicobacter pylori/efectos de los fármacos , Mycobacterium avium/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos
8.
ACS Infect Dis ; 5(6): 982-1000, 2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-30907573

RESUMEN

Nontuberculous mycobacteria (NTM) are ubiquitous microbes belonging to the Mycobacterium genus. Among all NTM pathogens, M. avium is one of the most frequent agents causing pulmonary disease, especially in immunocompromised individuals and cystic fibrosis patients. Recently, we reported the first ad hoc designed M. avium efflux pump inhibitor (EPI; 1b) able to strongly boost clarithromycin (CLA) MIC against different M. avium strains. Since the 3-phenylquinolone derivative 1b suffered from toxicity issues toward human macrophages, herein we report a two-pronged medicinal chemistry workflow for identifying new potent and safe NTM EPIs. Initially, we followed a computational approach exploiting our pharmacophore models to screen FDA approved drugs and in-house compounds to identify "ready-to-use" NTM EPIs and/or new scaffolds to be optimized in terms of EPI activity. Although nicardipine 2 was identified as a new NTM EPI, all identified molecules still suffered from toxicity issues. Therefore, based on the promising NTM EPI activity of 1b, we undertook the design, synthesis, and biological evaluation of new 3-phenylquinolones differently functionalized at the C6/C7 as well as N1 positions. Among the 27 synthesized 3-phenylquinolone analogues, compounds 11b, 12b, and 16a exerted excellent NTM EPI activity at concentrations below their CC50 on human cells, with derivative 16a being the most promising compound. Interestingly, 16a also showed good activity in M. avium-infected macrophages both alone as well as in combination with CLA. The antimycobacterial activity observed for 16a only when tested in the ex vivo model suggests a high therapeutic potential of EPIs against M. avium, which seems to need functional efflux pumps to establish intracellular infections.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Antibacterianos/farmacología , Benzoquinonas/química , Moduladores del Transporte de Membrana/farmacología , Proteínas de Transporte de Membrana/metabolismo , Mycobacterium/efectos de los fármacos , Simulación por Computador , Sinergismo Farmacológico , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Mycobacterium avium/efectos de los fármacos
9.
BMC Infect Dis ; 19(1): 203, 2019 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-30819109

RESUMEN

BACKGROUND: Nontuberculous mycobacterial (NTM) disease is commonly an opportunistic infection frequently found in immunocompromised individuals, but sometimes can also be found in the immunocompetent hosts, especially in East Asians. The NTM separation rate in China is increasing, which reminds us to focus on NTM infections in immunocompromised populations. CASE PRESENTATION: A 43-year-old woman with a recurrent fever for more than 8-month and a right forehead surgical wounds unhealed for more than 6-month was admitted to our hospital on February 22, 2018. On arrival, several elliptic ulcers were obvious on the right forehead with pus and fibrin exudation, and the skin around the lesions was tender, reddish, no sense of fluctuation. The result of HIV serology test was negative. CD4+ T cell count was normal and tuberculosis antibody was negative. CT of the chest and head showed bone destruction. Skin biopsy on the right forehead was performed on March 13, 2018, and pathological examination of the excisional biopsy specimen found inflammatory granuloma and suppurative inflammatory changes. Broad-spectrum antibiotics were treated but the effect seemed discontent. Then debridement and skin grafting were performed on the right frontal ulcer under general anesthesia on April 3, 2018. The skin tissue culture that resected on March 13, 2018 found Nontuberculous mycobacteria grown after 78 days, so clarithromycin, ethambutol, protionamide, and amoxicillin clavulanate potassium were prescribed for anti-nontuberculous mycobacteria treatment beginning on May 31, 2018. In reviewing the case, Mycobacterium avium (M. avium) was identified in the skin tissue resected on April 3, 2018 by polymerase chain reaction (PCR) and the serum test of anti-interferon-γ autoantibodies was positive. CONCLUSIONS: This is a case report of "Mycobacterium avium SSTI (skin and soft tissue infection) and OM (osteomyelitis) with possible secondary immunodeficiency syndrome induced by anti-interferon-γ autoantibody".


Asunto(s)
Interferón gamma/inmunología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium avium/patogenicidad , Osteomielitis/etiología , Infecciones de los Tejidos Blandos/tratamiento farmacológico , Adulto , Combinación Amoxicilina-Clavulanato de Potasio/uso terapéutico , Antibacterianos/uso terapéutico , Autoanticuerpos , Claritromicina/uso terapéutico , Etambutol/uso terapéutico , Femenino , Granuloma/microbiología , Granuloma/cirugía , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/genética , Osteomielitis/inmunología , Osteomielitis/terapia , Cuero Cabelludo/patología , Infecciones de los Tejidos Blandos/microbiología
10.
Int J Infect Dis ; 80: 134-136, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30639623

RESUMEN

Ruxolitinib is a JAK-1/JAK-2 inhibitor indicated for the treatment of polycythemia vera and primary or secondary myelofibrosis. Only one patient (0.2%) was diagnosed with tuberculosis among the 485 patients receiving ruxolitinib in the four pivotal trials. Fourteen cases of tuberculosis have since been reported. We observed two (3%) mycobacterial infections (one due to Mycobacterium tuberculosis and one due to Mycobacterium avium complex) in our cohort of 65 patients receiving ruxolitinib. This observation suggests that the rate of mycobacterial infection might be higher than that observed in the pivotal trials and that atypical mycobacterial infections can also occur.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/uso terapéutico , Tuberculosis/tratamiento farmacológico , Anciano , Femenino , Humanos , Masculino , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/aislamiento & purificación , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/aislamiento & purificación , Nitrilos , Policitemia Vera/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas
11.
Artículo en Inglés | MEDLINE | ID: mdl-30012773

RESUMEN

Nontuberculous mycobacteria (NTM) affect an increasing number of individuals worldwide. Infection with these organisms is more common in patients with chronic lung conditions, and treatment is challenging. Quinolones, such as ciprofloxacin, have been used to treat patients, but the results have not been encouraging. In this report, we evaluate novel formulations of liposome-encapsulated ciprofloxacin (liposomal ciprofloxacin) in vitro and in vivo Its efficacy against Mycobacterium avium and Mycobacterium abscessus was examined in macrophages, in biofilms, and in vivo using intranasal instillation mouse models. Liposomal ciprofloxacin was significantly more active than free ciprofloxacin against both pathogens in macrophages and biofilms. When evaluated in vivo, treatment with the liposomal ciprofloxacin formulations was associated with significant decreases in the bacterial loads in the lungs of animals infected with M. avium and M. abscessus In summary, topical delivery of liposomal ciprofloxacin in the lung at concentrations greater than those achieved in the serum can be effective in the treatment of NTM, and further evaluation is warranted.


Asunto(s)
Macrófagos/microbiología , Mycobacterium abscessus/efectos de los fármacos , Mycobacterium abscessus/patogenicidad , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/patogenicidad , Animales , Biopelículas/efectos de los fármacos , Femenino , Humanos , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Polietilenglicoles
12.
Mol Pharm ; 15(3): 1215-1225, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29421865

RESUMEN

The nontuberculous mycobacterial (NTM) pathogens, M. avium complex (MAC) and M. abscessus, can result in severe pulmonary infections. Current antibiotics confront significant challenges for treatment of these NTM infections due to emerging multidrug-resistance. Thus, development of new antibiotics targeted against these agents is needed. We examined the inhibitory activities of Ga(NO3)3, GaCl3, gallium meso-tetraphenylporphyrine (GaTP), and gallium nanoparticles (GaNP) against intra- and extracellular M. avium and M. abscessus. GaTP, an analogue of natural heme, inhibited growth of both M. avium and M. abscessus with MICs in Fe-free 7H9 media of 0.5 and 2 µg/mL, respectively. GaTP was more active than Ga(NO3)3 and GaCl3. Ga(NO3)3 and GaCl3 were not as active in Fe-rich media compared to Fe-free media. However, GaTP was much less impacted by exogenous Fe, with MICs against M. avium and M. abscessus of 2 and 4 µg/mL, respectively, in 7H9 OADC media (Fe rich). Confocal microscopy showed that GaNP penetrates the M. avium cell wall. As assessed by determining colony forming units, GaNP inhibited the growth of NTM growing in THP-1 macrophages up to 15 days after drug-loading of the cells, confirming a prolonged growth inhibitory activity of the GaNP. Biodistribution studies of GaNP conducted in mice showed that intraperitoneal injection is more effective than intramuscular injection in delivering Ga(III) into lung tissue. GaTP exhibits potential as a lead compound for development of anti-NTM agents that target heme-bound iron uptake mechanisms by mycobacteria and inhibit growth by disrupting mycobacterial iron acquisition/utilization.


Asunto(s)
Antibacterianos/farmacología , Galio/farmacología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Mycobacterium abscessus/efectos de los fármacos , Mycobacterium avium/efectos de los fármacos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Animales , Antibacterianos/uso terapéutico , Línea Celular Tumoral , Femenino , Galio/uso terapéutico , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Modelos Animales , Infecciones por Mycobacterium no Tuberculosas/microbiología , Nanopartículas/química , Porfirinas/química , Infecciones del Sistema Respiratorio/microbiología , Distribución Tisular
13.
Nanomedicine (Lond) ; 12(24): 2721-2736, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29119867

RESUMEN

AIM: This work aims to develop a mannosylated nanostructured lipid carrier (NLC) loaded with rifampicin to improve tuberculosis treatment. MATERIALS & METHODS: An active targeting strategy was used and the nanoparticles were characterized. Effects on cell viability and the antimycobacterial activity of the nanoformulations were evaluated. RESULTS: The nanoparticles developed exhibited a size of about 315 nm and polydispersity <0.2. The drug encapsulation efficiency was higher than 90% and its release was sensitive to pH. The mannosylated NLCs showed efficient uptake by bone marrow derived macrophages. Further, rifampicin-loaded mannosylated NLCs were more efficient in inducing a decrease of intracellular growth of mycobacteria. CONCLUSION: The NLCs developed can be used as a promising carrier for safer and efficient management of tuberculosis.


Asunto(s)
Antibióticos Antituberculosos/farmacología , Portadores de Fármacos/química , Lípidos/química , Macrófagos/efectos de los fármacos , Mycobacterium avium/efectos de los fármacos , Nanopartículas/química , Rifampin/farmacología , Tuberculosis Pulmonar/tratamiento farmacológico , Animales , Antibióticos Antituberculosos/química , Transporte Biológico , Supervivencia Celular , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Macrófagos/citología , Macrófagos/microbiología , Manosa/química , Ratones Endogámicos C57BL , Mycobacterium avium/aislamiento & purificación , Tamaño de la Partícula , Rifampin/química , Propiedades de Superficie
14.
Artículo en Inglés | MEDLINE | ID: mdl-29067283

RESUMEN

Mycobacterium tuberculosis (M.tb) infection manifests into tuberculosis (TB) in a small fraction of the infected population that comprises the TB susceptible group. Identifying the factors potentiating susceptibility to TB persistence is one of the prime agenda of TB control programs. Recently, WHO recognized diabetes as a risk factor for TB disease progression. The closely related pathological state of metabolic imbalance, dyslipidemia, is yet another emerging risk factor involving deregulation in host immune responses. While high cholesterol levels are clinically proven condition for perturbations in cardiac health, a significant fraction of population these days suffer from borderline risk cholesterol profiles. This apparently healthy population is susceptible to various health risks placing them in the "pre-disease" range. Our study focuses on determining the role of such asymptomatic dyslipidemia as a potential risk factor for susceptibility to TB persistence. Macrophages exposed to sub-pathological levels of cholesterol for chronic period, besides impaired release of TNF-α, could not clear intracellular pathogenic mycobacteria effectively as compared to the unexposed cells. These cells also allowed persistence of opportunistic mycobacterial infection by M. avium and M. bovis BCG, indicating highly compromised immune response. The cholesterol-treated macrophages developed a foamy phenotype with a significant increase in intracellular lipid-bodies prior to M.tb infection, potentially contributing to pre-disease state for tuberculosis infection. The foamy phenotype, known to support M.tb infection, increased several fold upon infection in these cells. Additionally, mitochondrial morphology and function were perturbed, more so during infection in cholesterol treated cells. Pharmacological supplementation with small molecule M1 that restored mitochondrial structural and functional integrity limited M.tb survival more effectively in cholesterol exposed macrophages. Mechanistically, M1 molecule promoted clearance of mycobacteria by reducing total cellular lipid content and restoring mitochondrial morphology and function to its steady state. We further supported our observations by infection assays in PBMC-derived macrophages from clinically healthy volunteers with borderline risk cholesterol profiles. With these observations, we propose that prolonged exposure to sub-pathological cholesterol can lead to asymptomatic susceptibility to M.tb persistence. Use of small molecules like M1 sets yet another strategy for host-directed therapy where re-functioning of mitochondria in cholesterol abused macrophages can improve M.tb clearance.


Asunto(s)
Antituberculosos/farmacología , Dislipidemias/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mycobacterium avium/efectos de los fármacos , Mycobacterium bovis/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/uso terapéutico , Colesterol/farmacología , Susceptibilidad a Enfermedades , Dislipidemias/inducido químicamente , Células Espumosas/inmunología , Humanos , Macrófagos/inmunología , Mitocondrias/fisiología , Cultivo Primario de Células , Factores de Riesgo , Células THP-1 , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Physiol Rep ; 4(21)2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27856731

RESUMEN

Mycobacterium avium (M. avium) causes significant pulmonary infection, especially in immunocompromised hosts. Alveolar macrophages (AMs) represent the first line of host defense against infection in the lung. Interferon gamma (IFN-γ) activation of AMs enhances in vitro killing of pathogens such as M. avium We hypothesized that airway delivery of AMs into the lungs of immunodeficient mice infected with M. avium will inhibit M. avium growth in the lung and that this macrophage function is in part IFN-γ dependent. In this study, normal BALB/c and BALB/c SCID mice received M. avium intratracheally while on mechanical ventilation. After 30 days, M. avium numbers increased in a concentration-dependent manner in SCID mice compared with normal BALB/c mice. Airway delivery of IFN-γ-activated BALB/c AMs or J774A.1 macrophages overexpressing IFN-γ into the lungs of SCID mice resulted in a significant decrease in M. avium growth (P < 0.01, both comparisons) and limited dissemination to other organs. In addition, airway delivery of IFN-γ activated AMs and macrophages overexpressing IFN-γ increased the levels of IFN-γ and TNF-α in SCID mice. A similar protective effect against M. avium infection using J774A.1 macrophages overexpressing IFN-γ was observed in IFN-γ knockout mice. These data suggest that administration of IFN-γ activated AMs or macrophages overexpressing IFN-γ may partially restore local alveolar host defense against infections like M. avium, even in the presence of ongoing systemic immunosuppression.


Asunto(s)
Interferón gamma/administración & dosificación , Pulmón/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones SCID/inmunología , Infecciones por Mycobacterium/inmunología , Mycobacterium avium/efectos de los fármacos , Animales , Tolerancia Inmunológica/efectos de los fármacos , Interferón gamma/biosíntesis , Interferón gamma/genética , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones por Mycobacterium/metabolismo , Infecciones por Mycobacterium/microbiología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
16.
BMC Microbiol ; 16(1): 251, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27793104

RESUMEN

BACKGROUND: Mycobacterium tuberculosis infection is thought to induce oxidative stress. N-acetyl-cysteine (NAC) is widely used in patients with chronic pulmonary diseases including tuberculosis due to its mucolytic and anti-oxidant activities. Here, we tested whether NAC exerts a direct antibiotic activity against mycobacteria. METHODS: Oxidative stress status in plasma was compared between pulmonary TB (PTB) patients and those with latent M. tuberculosis infection (LTBI) or healthy uninfected individuals. Lipid peroxidation, DNA oxidation and cell death, as well as accumulation of reactive oxygen species (ROS) were measured in cultures of primary human monocyte-derived macrophages infected with M. tuberculosis and treated or not with NAC. M. tuberculosis, M. avium and M. bovis BCG cultures were also exposed to different doses of NAC with or without medium pH adjustment to control for acidity. The anti-mycobacterial effect of NAC was assessed in M. tuberculosis infected human THP-1 cells and bone marrow-derived macrophages from mice lacking a fully functional NADPH oxidase system. The capacity of NAC to control M. tuberculosis infection was further tested in vivo in a mouse (C57BL/6) model. RESULTS: PTB patients exhibited elevated levels of oxidation products and a reduction of anti-oxidants compared with LTBI cases or uninfected controls. NAC treatment in M. tuberculosis-infected human macrophages resulted in a decrease of oxidative stress and cell death evoked by mycobacteria. Importantly, we observed a dose-dependent reduction in metabolic activity and in vitro growth of NAC treated M. tuberculosis, M. avium and M. bovis BCG. Furthermore, anti-mycobacterial activity in infected macrophages was shown to be independent of the effects of NAC on the host NADPH oxidase system in vitro. Short-term NAC treatment of M. tuberculosis infected mice in vivo resulted in a significant reduction of mycobacterial loads in the lungs. CONCLUSIONS: NAC exhibits potent anti-mycobacterial effects and may limit M. tuberculosis infection and disease both through suppression of the host oxidative response and through direct antimicrobial activity.


Asunto(s)
Acetilcisteína/farmacología , Antibacterianos/farmacología , Antioxidantes/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Muerte Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Humanos , Tuberculosis Latente/sangre , Tuberculosis Latente/tratamiento farmacológico , Tuberculosis Latente/microbiología , Peroxidación de Lípido/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/crecimiento & desarrollo , Mycobacterium avium/metabolismo , Mycobacterium bovis/efectos de los fármacos , Mycobacterium bovis/crecimiento & desarrollo , Mycobacterium bovis/metabolismo , NADPH Oxidasas/deficiencia , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología , Adulto Joven
17.
J Control Release ; 235: 112-124, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27261333

RESUMEN

Tuberculosis (TB), a disease caused by the human pathogen Mycobacterium tuberculosis, recently joined HIV/AIDS on the top rank of deadliest infectious diseases. Low patient compliance due to the expensive, long-lasting and multi-drug standard therapies often results in treatment failure and emergence of multi-drug resistant strains. In this scope, antimicrobial peptides (AMPs) arise as promising candidates for TB treatment. Here we describe the ability of the exogenous AMP LLKKK18 to efficiently kill mycobacteria. The peptide's potential was boosted by loading into self-assembling Hyaluronic Acid (HA) nanogels. These provide increased stability, reduced cytotoxicity and degradability, while potentiating peptide targeting to main sites of infection. The nanogels were effectively internalized by macrophages and the peptide presence and co-localization with mycobacteria within host cells was confirmed. This resulted in a significant reduction of the mycobacterial load in macrophages infected in vitro with the opportunistic M. avium or the pathogenic M. tuberculosis, an effect accompanied by lowered pro-inflammatory cytokine levels (IL-6 and TNF-α). Remarkably, intra-tracheal administration of peptide-loaded nanogels significantly reduced infection levels in mice infected with M. avium or M. tuberculosis, after just 5 or 10 every other day administrations. Considering the reported low probability of resistance acquisition, these findings suggest a great potential of LLKKK18-loaded nanogels for TB therapeutics.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/administración & dosificación , Antituberculosos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Geles/administración & dosificación , Ácido Hialurónico/administración & dosificación , Nanoestructuras/administración & dosificación , Animales , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Antituberculosos/química , Antituberculosos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Portadores de Fármacos/química , Portadores de Fármacos/uso terapéutico , Geles/química , Geles/uso terapéutico , Ácido Hialurónico/química , Ácido Hialurónico/uso terapéutico , Interleucina-6/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones Endogámicos C57BL , Mycobacterium avium/efectos de los fármacos , Mycobacterium avium/crecimiento & desarrollo , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/inmunología
18.
PLoS One ; 10(5): e0128772, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010725

RESUMEN

Mycobacterium avium subsp. hominissuis is an opportunistic pathogen that is associated with biofilm-related infections of the respiratory tract and is difficult to treat. In recent years, extracellular DNA (eDNA) has been found to be a major component of bacterial biofilms, including many pathogens involved in biofilm-associated infections. To date, eDNA has not been described as a component of mycobacterial biofilms. In this study, we identified and characterized eDNA in a high biofilm-producing strain of Mycobacterium avium subsp. hominissuis (MAH). In addition, we surveyed for presence of eDNA in various MAH strains and other nontuberculous mycobacteria. Biofilms of MAH A5 (high biofilm-producing strain) and MAH 104 (reference strain) were established at 22°C and 37°C on abiotic surfaces. Acellular biofilm matrix and supernatant from MAH A5 7 day-old biofilms both possess abundant eDNA, however very little eDNA was found in MAH 104 biofilms. A survey of MAH clinical isolates and other clinically relevant nontuberculous mycobacterial species revealed many species and strains that also produce eDNA. RAPD analysis demonstrated that eDNA resembles genomic DNA. Treatment with DNase I reduced the biomass of MAH A5 biofilms when added upon biofilm formation or to an already established biofilm both on abiotic surfaces and on top of human pharyngeal epithelial cells. Furthermore, co-treatment of an established biofilm with DNase 1 and either moxifloxacin or clarithromycin significantly increased the susceptibility of the bacteria within the biofilm to these clinically used antimicrobials. Collectively, our results describe an additional matrix component of mycobacterial biofilms and a potential new target to help treat biofilm-associated nontuberculous mycobacterial infections.


Asunto(s)
Biopelículas , ADN Bacteriano/fisiología , Farmacorresistencia Bacteriana , Mycobacterium avium/fisiología , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Línea Celular , Claritromicina/farmacología , Desoxirribonucleasa I/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Fluoroquinolonas/farmacología , Humanos , Moxifloxacino , Mycobacterium avium/efectos de los fármacos
19.
Molecules ; 19(7): 10386-409, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25036151

RESUMEN

In this study, a series of twenty-two ring-substituted naphthalene-1-carboxanilides were prepared and characterized. Primary in vitro screening of the synthesized carboxanilides was performed against Mycobacterium avium subsp. paratuberculosis. N-(2-Methoxyphenyl)naphthalene-1-carboxamide, N-(3-methoxy-phenyl)naphthalene-1-carboxamide, N-(3-methylphenyl)naphthalene-1-carboxamide, N-(4-methylphenyl)naphthalene-1-carboxamide and N-(3-fluorophenyl)naphthalene-1-carboxamide showed against M. avium subsp. paratuberculosis two-fold higher activity than rifampicin and three-fold higher activity than ciprofloxacin. The most effective antimycobacterial compounds demonstrated insignificant toxicity against the human monocytic leukemia THP-1 cell line. The testing of biological activity of the compounds was completed with the study of photosynthetic electron transport (PET) inhibition in isolated spinach (Spinacia oleracea L.) chloroplasts. The PET-inhibiting activity expressed by IC50 value of the most active compound N-[4-(trifluoromethyl)phenyl]naphthalene-1-carboxamide was 59 µmol/L. The structure-activity relationships are discussed.


Asunto(s)
Anilidas/química , Anilidas/farmacología , Naftalenos/química , Anilidas/síntesis química , Antibacterianos/química , Antibacterianos/farmacología , Cloroplastos/efectos de los fármacos , Cloroplastos/metabolismo , Transporte de Electrón/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Pruebas de Sensibilidad Microbiana , Mycobacterium avium/efectos de los fármacos , Fotosíntesis/efectos de los fármacos , Spinacia oleracea/efectos de los fármacos , Spinacia oleracea/metabolismo , Relación Estructura-Actividad
20.
Bioorg Med Chem ; 22(15): 4073-82, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24953953

RESUMEN

The development of novel antimicrobial drugs represents a cutting edge research topic. In this study, 20 salicylanilide N,N-disubstituted carbamates and thiocarbamates were designed, synthesised and characterised by IR, (1)H NMR and (13)C NMR. The compounds were evaluated in vitro as potential antimicrobial agents against Mycobacterium tuberculosis and nontuberculous mycobacteria (Mycobacterium avium and Mycobacterium kansasii) as well as against eight bacterial and fungal strains. Additionally, we investigated the inhibitory effect of these compounds on mycobacterial isocitrate lyase and cellular toxicity. The minimum inhibitory concentrations (MICs) against mycobacteria were from 4 µM for thiocarbamates and from 16 µM for carbamates. Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus, were inhibited with MICs from 0.49 µM by thiocarbamates, whilst Gram-negative bacteria and most of the fungi did not display any significant susceptibility. All (thio)carbamates mildly inhibited isocitrate lyase (up to 22%) at a concentration of 10 µM. The (thio)carbamoylation of the parent salicylanilides led to considerably decreased cytotoxicity and thus improved the selectivity indices (up to 175). These values indicate that some derivatives are attractive candidates for future research.


Asunto(s)
Antiinfecciosos/síntesis química , Carbamatos/química , Salicilanilidas/química , Tiocarbamatos/química , Antiinfecciosos/química , Antiinfecciosos/farmacología , Carbamatos/síntesis química , Carbamatos/farmacología , Supervivencia Celular/efectos de los fármacos , Hongos/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Células Hep G2 , Humanos , Isocitratoliasa/antagonistas & inhibidores , Isocitratoliasa/metabolismo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Mycobacterium avium/efectos de los fármacos , Mycobacterium kansasii/efectos de los fármacos , Relación Estructura-Actividad , Tiocarbamatos/síntesis química , Tiocarbamatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA